101
|
Lanzoni G, Linetsky E, Correa D, Alvarez RA, Marttos A, Hirani K, Cayetano SM, Castro JG, Paidas MJ, Efantis Potter J, Xu X, Glassberg M, Tan J, Patel AN, Goldstein B, Kenyon NS, Baidal D, Alejandro R, Vianna R, Ruiz P, Caplan AI, Ricordi C. Umbilical Cord-derived Mesenchymal Stem Cells for COVID-19 Patients with Acute Respiratory Distress Syndrome (ARDS). CELLR4-- REPAIR, REPLACEMENT, REGENERATION, & REPROGRAMMING 2020; 8. [PMID: 34164564 DOI: 10.32113/cellr4_20204_2839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The coronavirus SARS-CoV-2 is cause of a global pandemic of a pneumonia-like disease termed Coronavirus Disease 2019 (COVID-19). COVID-19 presents a high mortality rate, estimated at 3.4%. More than 1 out of 4 hospitalized COVID-19 patients require admission to an Intensive Care Unit (ICU) for respiratory support, and a large proportion of these ICU-COVID-19 patients, between 17% and 46%, have died. In these patients COVID-19 infection causes an inflammatory response in the lungs that can progress to inflammation with cytokine storm, Acute Lung Injury (ALI), Acute Respiratory Distress Syndrome (ARDS), thromboembolic events, disseminated intravascular coagulation, organ failure, and death. Mesenchymal Stem Cells (MSCs) are potent immunomodulatory cells that recognize sites of injury, limit effector T cell reactions, and positively modulate regulatory cell populations. MSCs also stimulate local tissue regeneration via paracrine effects inducing angiogenic, anti-fibrotic and remodeling responses. MSCs can be derived in large number from the Umbilical Cord (UC). UC-MSCs, utilized in the allogeneic setting, have demonstrated safety and efficacy in clinical trials for a number of disease conditions including inflammatory and immune-based diseases. UC-MSCs have been shown to inhibit inflammation and fibrosis in the lungs and have been utilized to treat patients with severe COVID-19 in pilot, uncontrolled clinical trials, that reported promising results. UC-MSCs processed at our facility have been authorized by the FDA for clinical trials in patients with an Alzheimer's Disease, and in patients with Type 1 Diabetes (T1D). We hypothesize that UC-MSC will also exert beneficial therapeutic effects in COVID-19 patients with cytokine storm and ARDS. We propose an early phase controlled, randomized clinical trial in COVID-19 patients with ALI/ARDS. Subjects in the treatment group will be treated with two doses of UC-MSC (l00 × 106 cells). The first dose will be infused within 24 hours following study enrollment. A second dose will be administered 72 ± 6 hours after the first infusion. Subject in the control group will receive infusion of vehicle (DPBS supplemented with 1% HSA and 70 U/kg unfractionated Heparin, delivered IV) following the same timeline. Subjects will be evaluated daily during the first 6 days, then at 14, 28, 60, and 90 days following enrollment (see Schedule of Assessment for time window details). Safety will be determined by adverse events (AEs) and serious adverse events (SAEs) during the follow-up period. Efficacy will be defined by clinical outcomes, as well as a variety of pulmonary, biochemical and immunological tests. Success of the current study will provide a framework for larger controlled, randomized clinical trials and a means of accelerating a possible solution for this urgent but unmet medical need. The proposed early phase clinical trial will be performed at the University of Miami (UM), in the facilities of the Diabetes Research Institute (DRI), UHealth Intensive Care Unit (ICU) and the Clinical Translational Research Site (CTRS) at the University of Miami Miller School of Medicine and at the Jackson Memorial Hospital (JMH).
Collapse
Affiliation(s)
- G Lanzoni
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - E Linetsky
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - D Correa
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Orthopedics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - R A Alvarez
- University of Miami Health System and Jackson Health System, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - A Marttos
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,University of Miami Health System and Jackson Health System, Miami, FL, USA
| | - K Hirani
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - S Messinger Cayetano
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - J G Castro
- University of Miami Health System and Jackson Health System, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - M J Paidas
- University of Miami Health System and Jackson Health System, Miami, FL, USA.,Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - J Efantis Potter
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - X Xu
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M Glassberg
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - J Tan
- Organ Transplant Institute, Fuzhou General Hospital, Xiamen University, Fuzhou, China
| | - A N Patel
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.,HCA Research Institute, Nashville, TN, USA
| | - B Goldstein
- Department of Head and Neck Surgery and Communication Sciences, Duke University, Durham, NC, USA
| | - N S Kenyon
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - D Baidal
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - R Alejandro
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - R Vianna
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,University of Miami Health System and Jackson Health System, Miami, FL, USA.,Miami Transplant Institute, Jackson Health System, Miami, FL, USA
| | - P Ruiz
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,University of Miami Health System and Jackson Health System, Miami, FL, USA.,Miami Transplant Institute, Jackson Health System, Miami, FL, USA
| | - A I Caplan
- Department of Medicine, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - C Ricordi
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,University of Miami Health System and Jackson Health System, Miami, FL, USA
| |
Collapse
|
102
|
Abstract
Mesenchymal stromal or stem cells (MSC) possess strong immunomodulatory properties. Due to their impressive potential to differentiate into various cell types they are capable of inducing mechanisms of tissue repair. Experimental data have demonstrated impaired MSC function in several rheumatic diseases in vitro; however, the relevance of these phenomena for the pathogenesis of rheumatic disorders has not been convincingly demonstrated. Nevertheless, allogeneic MSC transplantation (MSCT), and possibly autologous MSCT as well, could prove to be an interesting instrument for the treatment of autoimmune rheumatic diseases. The first clinical trials have demonstrated positive effects in systemic lupus erythematosus, systemic sclerosis and Sjogren's syndrome; however, questions regarding the long-term benefits and safety as well as the best source, the optimal cultivation technique and the most effective way of application of MSC are still unanswered.
Collapse
|
103
|
Zhou X, Jin N, Wang F, Chen B. Mesenchymal stem cells: a promising way in therapies of graft-versus-host disease. Cancer Cell Int 2020; 20:114. [PMID: 32280306 PMCID: PMC7137413 DOI: 10.1186/s12935-020-01193-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
It is well acknowledged that allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an effective treatment for numerous malignant blood diseases, which has also been applied to autoimmune diseases for more than a decade. Whereas graft-versus-host disease (GVHD) occurs after allogeneic hematopoietic stem cell transplantation (allo-HSCT) as a common serious complication, seriously affecting the efficacy of transplantation. Mesenchymal stem cells (MSCs) derived from a wealth of sources can easily isolate and expand with low immunogenicity. MSCs also have paracrine and immune regulatory functions, leading to a broad application prospect in treatment and tissue engineering. This review focuses on immunoregulatory function of MSCs, factors affecting mesenchymal stem cells to exert immunosuppressive effects, clinical application of MSCs in GVHD and researches on MSC-derived extracellular vesicles (EVs). The latest research progress on MSC in related fields is reviewed as well. The relevant literature from PubMed databases is reviewed in this article.
Collapse
Affiliation(s)
- Xinyi Zhou
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009 Jiangsu People's Republic of China
| | - Nan Jin
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009 Jiangsu People's Republic of China
| | - Fei Wang
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009 Jiangsu People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009 Jiangsu People's Republic of China
| |
Collapse
|
104
|
Chu L, Luo Y, Chen H, Miao Q, Wang L, Moats R, Wang T, Kennedy JC, Henske EP, Shi W. Mesenchymal folliculin is required for alveolar development: implications for cystic lung disease in Birt-Hogg-Dubé syndrome. Thorax 2020; 75:486-493. [PMID: 32238524 DOI: 10.1136/thoraxjnl-2019-214112] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/13/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Pulmonary cysts and spontaneous pneumothorax are presented in most patients with Birt-Hogg-Dubé (BHD) syndrome, which is caused by loss of function mutations in the folliculin (FLCN) gene. The pathogenic mechanisms underlying the cystic lung disease in BHD are poorly understood. METHODS Mesenchymal Flcn was specifically deleted in mice or in cultured lung mesenchymal progenitor cells using a Cre/loxP approach. Dynamic changes in lung structure, cellular and molecular phenotypes and signalling were measured by histology, immunofluorescence staining and immunoblotting. RESULTS Deletion of Flcn in mesoderm-derived mesenchymal cells results in significant reduction of postnatal alveolar growth and subsequent alveolar destruction, leading to cystic lesions. Cell proliferation and alveolar myofibroblast differentiation are inhibited in the Flcn knockout lungs, and expression of the extracellular matrix proteins Col3a1 and elastin are downregulated. Signalling pathways including mTORC1, AMP-activated protein kinase, ERK1/2 and Wnt-β-catenin are differentially affected at different developmental stages. All the above changes have statistical significance (p<0.05). CONCLUSIONS Mesenchymal Flcn is an essential regulator during alveolar development and maintenance, through multiple cellular and molecular mechanisms. The mesenchymal Flcn knockout mouse model provides the first in vivo disease model that may recapitulate the stages of cyst development in human BHD. These findings elucidate the developmental origins and mechanisms of lung disease in BHD.
Collapse
Affiliation(s)
- Ling Chu
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yongfeng Luo
- The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Hui Chen
- The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Qing Miao
- The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Larry Wang
- The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Rex Moats
- The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Tiansheng Wang
- The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - John C Kennedy
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Elizabeth P Henske
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wei Shi
- The Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
105
|
Yoshida S, Miyagawa S, Toyofuku T, Fukushima S, Kawamura T, Kawamura A, Kashiyama N, Nakamura Y, Toda K, Sawa Y. Syngeneic Mesenchymal Stem Cells Reduce Immune Rejection After Induced Pluripotent Stem Cell-Derived Allogeneic Cardiomyocyte Transplantation. Sci Rep 2020; 10:4593. [PMID: 32165680 PMCID: PMC7067786 DOI: 10.1038/s41598-020-58126-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/07/2020] [Indexed: 01/01/2023] Open
Abstract
Avoiding immune rejection after allogeneic induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) transplantation is a concern. However, mesenchymal stem cells (MSCs) can suppress immune rejection. To determine whether MSC co-transplantation can reduce immune rejection after allogeneic iPSC-CM transplantation, the latter cell type, harbouring a luciferase transgene, was subcutaneously transplanted alone or together with syngeneic MSCs into BALB/c mice. Bioluminescence imaging revealed that MSC co-transplantation significantly improved graft survival (day 7: iPSC-CMs alone 34 ± 5%; iPSC-CMs with MSCs, 61 ± 7%; P = 0.008). MSC co-transplantation increased CD4 + CD25 + FOXP3 + regulatory T cell numbers, apoptotic CD8-positive T cells, and IL-10 and TGF-beta expression at the implantation site. Analysis using a regulatory T cell depletion model indicated that enhanced regulatory T cell populations in the iPSC-CM with MSC group partially contributed to the extended iPSC-CM survival. Further, MSCs affected activated lymphocytes directly through cell–cell contact, which reduced the CD8/CD4 ratio, the proportion of Th1-positive cells among CD4-positive cells, and the secretion of several inflammation-related cytokines. Syngeneic MSC co-transplantation might thus control allogeneic iPSC-CM rejection by mediating immune tolerance via regulatory T cells and cell–cell contact with activated lymphocytes; this approach has promise for cardiomyogenesis-based therapy using allogeneic iPSC-CMs for severe heart failure.
Collapse
Affiliation(s)
- Shohei Yoshida
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshihiko Toyofuku
- Department of Immunology and Molecular Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Satsuki Fukushima
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Ai Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Kashiyama
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuki Nakamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Koichi Toda
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
106
|
Zhou T, Liao C, Li HY, Lin W, Lin S, Zhong H. Efficacy of mesenchymal stem cells in animal models of lupus nephritis: a meta-analysis. Stem Cell Res Ther 2020; 11:48. [PMID: 32019582 PMCID: PMC7001209 DOI: 10.1186/s13287-019-1538-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/11/2019] [Accepted: 12/20/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Lupus nephritis is usually manifested by proteinuria, active urinary sediment, hypertension, and renal failure and is a serious complication with more than 50% occurrence in systemic lupus erythematosus patients. Mesenchymal stem cells (MSC) present remarkable immunomodulatory ability, and these cells are potential therapeutic agents for autoimmune disorders. In clinical trials, the effectiveness of MSC in the treatment of lupus nephritis is still controversial. A meta-analysis was performed to assess whether MSC can achieve good efficacy in the treatment of lupus nephritis in mice. METHODS A comprehensive literature search was performed in Cochrane Library, ISI Web of Science, PubMed, and EMBASE from inception to Oct 1, 2019. Two authors independently extracted the data, which were pooled and calculated using RevMan 5.3. RESULTS A total of 28 studies met the inclusion criteria. MSC treatment resulted in lower levels of ds-DNA (OR = - 29.58, 95% CI - 29.58, - 17.99; P < 0.00001), ANA (OR = - 70.93, 95% CI - 104.55, - 37.32; P < 0.0001), Scr (OR = - 8.20, 95% CI - 12.71, - 3.69; P = 0.0004), BUN (OR = - 14.57, 95% CI - 20.50, - 8.64; P < 0.00001), proteinuria (OR = - 4.26, 95% CI - 5.15 to - 3.37; P < 0.00001), and renal sclerosis score (OR = - 1.92, 95% CI - 2.66 to - 1.18; P < 0.00001), and MSC treatment could get higher levels of albumin. To detect the potential, the cytokines were also assessed, and the MSC treatment group had lower levels of IL-2, IL-12, IL-17, and IFN-γ when compared with the control group. However, the difference was not notable for IL-4, IL-6, IL-10, TGF-β, MCP-1, TNF-α, Th1, Th17, Foxp3, or Tregs. CONCLUSION Our study confirmed that MSC treatment in an animal model for lupus nephritis in the studies included in the meta-analysis resulted in lower levels of ds-DNA, ANA, Scr, BUN, proteinuria, and renal sclerosis score, and MSC treatment could get higher levels of albumin.
Collapse
Affiliation(s)
- Tianbiao Zhou
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, 515041, No. 69 Dongsha Road, Shantou, China.
| | - Chunling Liao
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, 515041, No. 69 Dongsha Road, Shantou, China
| | - Hong-Yan Li
- Department of Nephrology, Huadu District People's Hospital of Guangzhou, Southern Medical University, Guangzhou, 510800, China
| | - Wenshan Lin
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, 515041, No. 69 Dongsha Road, Shantou, China
| | - Shujun Lin
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, 515041, No. 69 Dongsha Road, Shantou, China
| | - Hongzhen Zhong
- Department of Nephrology, the Second Affiliated Hospital, Shantou University Medical College, 515041, No. 69 Dongsha Road, Shantou, China
| |
Collapse
|
107
|
Wu KH, Cheng CC, Li JP, Weng TF, Yang SF, Pan HH, Chao YH. Toll-like receptor signalling associated with immunomodulation of umbilical cord-derived mesenchymal stem cells in mice with systemic lupus erythematosus. Lupus 2020; 29:165-175. [PMID: 31964222 DOI: 10.1177/0961203319898532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
With potent immunomodulatory activities, mesenchymal stem cells (MSCs) have the potential to be a beneficial treatment option for diseases with aberrant immune responses such as systemic lupus erythematosus (SLE). However, the underlying mechanisms remain largely unknown. Here, we used NZBWF1 mice as a SLE animal model to examine immunomodulation of MSCs as well as to assess the role of Toll-like receptor signalling in this circumstance. We found that mice receiving MSCs had a significant decrease in severity of proteinuria at 20 and 22 weeks of age (p = 0.009 and p = 0.022, respectively). Serum anti-dsDNA levels were significantly lower compared with the control group (p = 0.016 and p = 0.036, respectively). C3 and C4 levels were significantly higher at 22 weeks of age (p = 0.046 and p = 0.016, respectively). Altered expression of inflammation-associated cytokine profiles in the serum was also noted in mice receiving MSCs. Down-regulation of myeloid differentiation factor 88 (MyD88)-nuclear factor-κB (NF-κB) signalling in the liver was demonstrated by quantitative polymerase chain reaction, ELISA and Western blotting. In addition to demonstrating the beneficial effects of MSC treatment in NZBWF1 mice, our study provided the first evidence for the association of MyD88-NF-κB signalling and MSC-mediated immunomodulation in this disease.
Collapse
Affiliation(s)
- K H Wu
- Division of Pediatric Hematology-Oncology, Children's Hospital, China Medical University, Taichung, Taiwan.,School of Post-baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
| | - C C Cheng
- Laboratory Animal Service Center, Office of Research and Development, China Medical University, Taichung, Taiwan
| | - J P Li
- Rheumatology Research Center, China Medical University Hospital, Taichung, Taiwan.,School of Dentistry, National Defense Medical Center, Taipei, Taiwan
| | - T F Weng
- Division of Pediatric Hematology-Oncology, Children's Hospital, China Medical University, Taichung, Taiwan
| | - S F Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - H H Pan
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Y H Chao
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
108
|
Deniz AAH, Abdik EA, Abdik H, Aydın S, Şahin F, Taşlı PN. Zooming in across the Skin: A Macro-to-Molecular Panorama. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1247:157-200. [DOI: 10.1007/5584_2019_442] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
109
|
Effects of Human Amnion-Derived Mesenchymal Stem Cell (hAD-MSC) Transplantation In Situ on Primary Ovarian Insufficiency in SD Rats. Reprod Sci 2020; 27:1502-1512. [PMID: 31953773 DOI: 10.1007/s43032-020-00147-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022]
Abstract
Human amnion-derived mesenchymal stem cell (hAD-MSC) transplantation can repair ovarian injury and improve ovarian function in rats with chemotherapy-induced primary ovarian insufficiency (POI). However, ensuring that stem cells home to the ovary to improve their effects on ovarian injury is challenging. This research aimed to directly inject ovarian tissue with hAD-MSCs and improve the homing of stem cells to the ovary. The animals were divided into POI, hAD-MSC (tail vein) treatment, hAD-MSC (in situ) treatment, and control groups. POI rat models were established by intraperitoneal injection of cyclophosphamide (CTX) and busulfan (BUS). The hAD-MSCs isolated from the amnion were injected into the tail vein or ovary of POI rats. The estrous cycle, serum sex hormone levels, follicle counts, ovarian pathological changes, and proteome of the ovaries were evaluated. hAD-MSCs were successfully isolated and cultured from the amnion. Both hAD-MSC (tail vein) and hAD-MSC (in situ) transplantation increased body weight, improved the AMH levels and follicle numbers, and reduced reproductive organ injuries in POI rats. Transplantation of hAD-MSCs (in situ) upregulated 24 proteins and downregulated 4 proteins. Both hAD-MSC (tail vein) and hAD-MSC (in situ) transplantations can repair ovarian injury and improve ovarian function in rats with chemotherapy-induced POI. The paracrine proteome of hAD-MSCs in the ovarian microenvironment can protect against chemotherapy-induced damage by reducing apoptosis and promoting angiogenesis, cell proliferation, and gene expression.
Collapse
|
110
|
Li H, Tian Y, Xie L, Liu X, Huang Z, Su W. Mesenchymal stem cells in allergic diseases: Current status. Allergol Int 2020; 69:35-45. [PMID: 31445840 DOI: 10.1016/j.alit.2019.08.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/17/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022] Open
Abstract
Allergic diseases, which include asthma, allergic skin diseases, allergic rhinitis and allergic conjunctivitis, have already garnered worldwide public health attention over recent decades. Mesenchymal stem cells (MSCs) have gradually emerged as a potential method for treating allergic diseases due to their immunosuppressive characteristics, tissue repair ability and secretion of various biological factors. This potential of MSC-based therapy has been confirmed in clinical and preclinical studies, which report the therapeutic benefits of MSCs for various allergic diseases and explore the antiallergic mechanisms. In this review, we focus on the discoveries and biological mechanisms of MSCs as a therapeutic tool in allergic diseases. We discuss the challenges of conducting MSC studies as well as future directions.
Collapse
|
111
|
Yu T, Yan B, Li J, Zhang T, Yang R, Wang X, Liu Y, Liu D. Acetylsalicylic acid rescues the immunomodulation of inflamed gingiva-derived mesenchymal stem cells via upregulating FasL in mice. Stem Cell Res Ther 2019; 10:368. [PMID: 31796122 PMCID: PMC6892130 DOI: 10.1186/s13287-019-1485-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 02/06/2023] Open
Abstract
Background Gingiva-derived mesenchymal stem cells (GMSCs) obtained multipotent differentiation and immunomodulatory properties. However, collecting healthy gingival tissues may be challenging in the clinical situation. Thus, in our present study, we aim to evaluate whether the immunomodulatory capacity of gingiva-derived mesenchymal stem cells from inflamed gingival tissues (iGMSCs) is impaired and find a way to rescue their deficient properties. Methods We compared the immunomodulation capacity of GMSCs and iGMSCs using an in vitro co-culture system and a mouse colitis model. T cell apoptosis, T helper 17 (Th17), and regulatory T (Treg) cell differentiation were detected by flow cytometry analysis. Results We demonstrated that iGMSCs obtained a decreased immunomodulatory capacity compared with GMSCs. Acetylsalicylic acid (ASA) pretreatment was able to rescue iGMSCs’ impaired immunomodulatory properties. Mechanistically, ASA was capable of upregulating the expression of Fas ligand (FasL) in iGMSCs, leading to an improvement in iGMSC-mediated T cell apoptosis and therapeutic efficacy in the treatment in colitis mice. Conclusions This study indicates that the deficient immunomodulatory function of iGMSCs could be rescued by ASA pretreatment via upregulating of FasL in mice. This strategy might serve as a practical approach to rescue deficient MSC function for further therapeutic application.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Orthodontics, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Boxi Yan
- Second Clinical Division, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Jing Li
- Department of Orthodontics, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Ting Zhang
- Department of Orthodontics, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Ruili Yang
- Department of Orthodontics, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Xuedong Wang
- Department of Orthodontics, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Yan Liu
- Department of Orthodontics, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China
| | - Dawei Liu
- Department of Orthodontics, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, People's Republic of China.
| |
Collapse
|
112
|
Short and Long Term Clinical and Immunologic Follow up after Bone Marrow Mesenchymal Stromal Cell Therapy in Progressive Multiple Sclerosis-A Phase I Study. J Clin Med 2019; 8:jcm8122102. [PMID: 31810187 PMCID: PMC6947442 DOI: 10.3390/jcm8122102] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow derived mesenchymal stromal cells (BM-MSCs) have emerged as a possible new therapy for Multiple Sclerosis (MS), however studies regarding efficacy and in vivo immune response have been limited and inconclusive. We conducted a phase I clinical study assessing safety and clinical and peripheral immune responses after MSC therapy in MS. Seven patients with progressive MS were intravenously infused with a single dose of autologous MSC (1–2 × 106 MSCs/kg body weight). The infusions were safe and well tolerated when given during clinical remission. Five out of seven patients completed the follow up of 48 weeks post-infusion. Brain magnetic resonance imaging (MRI) showed the absence of new T2 lesions at 12 weeks in 5/6 patients, while 3/5 had accumulated new T2 lesions at 48 weeks. Patient expanded disability status scales (EDSS) were stable in 6/6 at 12 weeks but declined in 3/5 patients at 48 weeks. Early changes of circulating microRNA levels (2 h) and increased proportion of FOXP3+ Tregs were detected at 7 days post-infusion compared to baseline levels. In conclusion, MSC therapy was safe and well tolerated and is associated with possible transient beneficial clinical and peripheral immunotolerogenic effects.
Collapse
|
113
|
Treatment with adipose tissue-derived mesenchymal stem cells exerts anti-diabetic effects, improves long-term complications, and attenuates inflammation in type 2 diabetic rats. Stem Cell Res Ther 2019; 10:333. [PMID: 31747961 PMCID: PMC6868748 DOI: 10.1186/s13287-019-1474-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/03/2019] [Accepted: 10/30/2019] [Indexed: 02/08/2023] Open
Abstract
Background Long-term diabetes-associated complications are the major causes of morbidity and mortality in individuals with diabetes. These diabetic complications are closely linked to immune system activation along with chronic, non-resolving inflammation, but therapies to directly reverse these complications are still not available. Our previous study demonstrated that mesenchymal stem cells (MSCs) attenuated chronic inflammation in type 2 diabetes mellitus (T2DM), resulting in improved insulin sensitivity and islet function. Therefore, we speculated that MSCs might exert anti-inflammatory effects and promote the reversal of diabetes-induced kidney, liver, lung, heart, and lens diseases in T2DM rats. Methods We induced a long-term T2DM complication rat model by using a combination of a low dose of streptozotocin (STZ) with a high-fat diet (HFD) for 32 weeks. Adipose-derived mesenchymal stem cells (ADSCs) were systemically administered once a week for 24 weeks. Then, we investigated the role of ADSCs in modulating the progress of long-term diabetic complications. Results Multiple infusions of ADSCs attenuated chronic kidney disease (CKD), nonalcoholic steatohepatitis (NASH), lung fibrosis, and cataracts; improved cardiac function; and lowered serum lipid levels in T2DM rats. Moreover, the levels of inflammatory cytokines in the serum of each animal group revealed that ADSC infusions were able to not only inhibit pro-inflammatory cytokines IL-6, IL-1β, and TNF-α expression but also increase anti-inflammatory cytokine IL-10 systematically. Additionally, MSCs reduced the number of iNOS(+) M1 macrophages and restored the number of CD163(+) M2 macrophages. Conclusions Multiple intravenous infusions of ADSCs produced significant protective effects against long-term T2DM complications by alleviating inflammation and promoting tissue repair. The present study suggests ADSCs may be a novel, alternative cell therapy for long-term diabetic complications.
Collapse
|
114
|
Cheng RJ, Xiong AJ, Li YH, Pan SY, Zhang QP, Zhao Y, Liu Y, Marion TN. Mesenchymal Stem Cells: Allogeneic MSC May Be Immunosuppressive but Autologous MSC Are Dysfunctional in Lupus Patients. Front Cell Dev Biol 2019; 7:285. [PMID: 31799252 PMCID: PMC6874144 DOI: 10.3389/fcell.2019.00285] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/04/2019] [Indexed: 02/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have a potently immunosuppressive capacity in both innate and adaptive immune responses. Consequently, MSCs transplantation has emerged as a potential beneficial therapy for autoimmune diseases even though the mechanisms underlying the immunomodulatory activity of MSCs is incompletely understood. Transplanted MSCs from healthy individuals with no known history of autoimmune disease are immunosuppressive in systemic lupus erythematosus (SLE) patients and can ameliorate SLE disease symptoms in those same patients. In contrast, autologous MSCs from SLE patients are not immunosuppressive and do not ameliorate disease symptoms. Recent studies have shown that MSCs from SLE patients are dysfunctional in both proliferation and immunoregulation and phenotypically senescent. The senescent phenotype has been attributed to multiple genes and signaling pathways. In this review, we focus on the possible mechanisms for the defective phenotype and function of MSCs from SLE patients and summarize recent research on MSCs in autoimmune diseases.
Collapse
Affiliation(s)
- Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - An-Ji Xiong
- Department of Rheumatology and Immunology, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Yan-Hong Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Shu-Yue Pan
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Qiu-Ping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Tony N Marion
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
115
|
Xu J, Chen J, Li W, Lian W, Huang J, Lai B, Li L, Huang Z. Additive Therapeutic Effects of Mesenchymal Stem Cells and IL-37 for Systemic Lupus Erythematosus. J Am Soc Nephrol 2019; 31:54-65. [PMID: 31604808 DOI: 10.1681/asn.2019050545] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 09/11/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although mesenchymal stem cells (MSCs) might offer a promising strategy for treating SLE, their immunoregulatory plasticity makes their therapeutic effects unpredictable. Whether overexpressing IL-37, an IL-1 family member with immunosuppressive activity, might enhance the therapeutic effects of these cells for SLE is unknown. METHODS We genetically modified MSCs to overexpress IL-37 and assessed their effects on immune suppression in vitro. We also evaluated the effects of such cells versus effects of various controls after transplanting them into MRL/lpr mice (model of SLE). RESULTS Stem cell characteristics did not appear altered in MSCs overexpressing IL-37. These cells had enhanced immunosuppression in vitro in terms of inhibiting splenocyte proliferation, reducing proinflammatory factors (IL-1β, TNF-α, IL-17, and IL-6), and suppressing autoantibodies (anti-dsDNA and anti-ANA). Compared with animals receiving control MSCs or IL-37 treatment alone, MRL/lpr mice transplanted with IL-37-overexpressing cells displayed improved survival and reduced signs of SLE (indicated by urine protein levels, spleen weight, and renal pathologic scores); they also had significantly lower expression of proinflammatory factors, lower total antibody levels in serum and urine, lower autoantibody production, and showed reduced T cell numbers in the serum and kidney. Expression of IL-37 by MSCs can maintain higher serum levels of IL-37, and MSCs had prolonged survival after transplantation, perhaps through IL-37 suppressing the inflammatory microenvironment. CONCLUSIONS Mutually reinforcing interaction between MSCs and IL-37 appears to underlie their additive therapeutic effects. Genetic modification to overexpress IL-37 might offer a way to enhance the stability and effectiveness of MSCs in treating SLE.
Collapse
Affiliation(s)
- Jianyong Xu
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease and .,Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, P.R. China
| | - Jieting Chen
- Department of Obstetrics, People's Hospital of Baoan, Shenzhen, P.R. China; and
| | - Wenlei Li
- Department of Obstetrics, Women and Children Health Institute of Futian, Shenzhen, P.R. China
| | - Wei Lian
- Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, P.R. China
| | - Jieyong Huang
- Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, P.R. China
| | - Baoyu Lai
- Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, P.R. China
| | - Lingyun Li
- Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, P.R. China
| | - Zhong Huang
- Guangdong Provincial Key Laboratory of Regional Immunity and Disease and .,Department of Immunology, Health Science Center, Shenzhen University, Shenzhen, P.R. China
| |
Collapse
|
116
|
The Effects of Hypoxia on the Immune-Modulatory Properties of Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells Int 2019; 2019:2509606. [PMID: 31687031 PMCID: PMC6800910 DOI: 10.1155/2019/2509606] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 08/11/2019] [Accepted: 09/09/2019] [Indexed: 01/09/2023] Open
Abstract
The therapeutic repertoire for life-threatening inflammatory conditions like sepsis, graft-versus-host reactions, or colitis is very limited in current clinical practice and, together with chronic ones, like the osteoarthritis, presents growing economic burden in developed countries. This urges the development of more efficient therapeutic modalities like the mesenchymal stem cell-based approaches. Despite the encouraging in vivo data, however, clinical trials delivered ambiguous results. Since one of the typical features of inflamed tissues is decreased oxygenation, the success of cellular therapy in inflammatory pathologies seems to be affected by the impact of oxygen depletion on transplanted cells. Here, we examine our current knowledge on the effect of hypoxia on the physiology of bone marrow-derived mesenchymal stromal cells, one of the most popular tools of practical cellular therapy, in the context of their immune-modulatory capacity.
Collapse
|
117
|
Tan W, Gu Z, Leng J, Zou X, Chen H, Min F, zhou W, Zhang L, Li G. Let-7f-5p ameliorates inflammation by targeting NLRP3 in bone marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Biomed Pharmacother 2019; 118:109313. [DOI: 10.1016/j.biopha.2019.109313] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
|
118
|
Abughanam G, Elkashty OA, Liu Y, Bakkar MO, Tran SD. Mesenchymal Stem Cells Extract (MSCsE)-Based Therapy Alleviates Xerostomia and Keratoconjunctivitis Sicca in Sjogren's Syndrome-Like Disease. Int J Mol Sci 2019; 20:ijms20194750. [PMID: 31557796 PMCID: PMC6801785 DOI: 10.3390/ijms20194750] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/16/2022] Open
Abstract
Sjogren’s syndrome (SS) is an autoimmune disease that manifests primarily in salivary and lacrimal glands leading to dry mouth and eyes. Unfortunately, there is no cure for SS due to its complex etiopathogenesis. Mesenchymal stem cells (MSCs) were successfully tested for SS, but some risks and limitations remained for their clinical use. This study combined cell- and biologic-based therapies by utilizing the MSCs extract (MSCsE) to treat SS-like disease in NOD mice. We found that MSCsE and MSCs therapies were successful and comparable in preserving salivary and lacrimal glands function in NOD mice when compared to control group. Cells positive for AQP5, AQP4, α-SMA, CK5, and c-Kit were preserved. Gene expression of AQP5, EGF, FGF2, BMP7, LYZ1 and IL-10 were upregulated, and downregulated for TNF-α, TGF-β1, MMP2, CASP3, and IL-1β. The proliferation rate of the glands and serum levels of EGF were also higher. Cornea integrity and epithelial thickness were maintained due to tear flow rate preservation. Peripheral tolerance was re-established, as indicated by lower lymphocytic infiltration and anti-SS-A antibodies, less BAFF secretion, higher serum IL-10 levels and FoxP3+ Treg cells, and selective inhibition of B220+ B cells. These promising results opened new venues for a safer and more convenient combined biologic- and cell-based therapy.
Collapse
Affiliation(s)
- Ghada Abughanam
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7, Canada.
| | - Osama A Elkashty
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7, Canada.
| | - Younan Liu
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7, Canada.
| | - Mohammed O Bakkar
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7, Canada.
| | - Simon D Tran
- McGill Craniofacial Tissue Engineering and Stem Cells Laboratory, Faculty of Dentistry, McGill University, Montreal, QC H3A 0C7, Canada.
| |
Collapse
|
119
|
Xu J, Su Y, Hu L, Cain A, Gu Y, Liu B, Wu R, Wang S, Wang H. Effect of Bone Morphogenetic Protein 6 on Immunomodulatory Functions of Salivary Gland-Derived Mesenchymal Stem Cells in Sjögren's Syndrome. Stem Cells Dev 2019; 27:1540-1548. [PMID: 30132383 DOI: 10.1089/scd.2017.0161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sjögren's syndrome (SS) is characterized by autoimmune activation and loss of function in the salivary glands. Recent studies reported that bone morphogenetic protein 6 (BMP6), which is a member of transforming growth factor beta (TGF-β) superfamily, was highly expressed in SS patients. To investigate the role of BMP6 in SS, we treated the salivary gland-derived mesenchymal stem cells (SGMSCs) with BMP6 and found that BMP6 could impair immunomodulatory properties of normal SGMSCs by downregulating the Prostaglandin E2 synthase through DNA-binding protein inhibitor-1. Neutralizing the BMP6 could significantly restore the SGMSC's immunoregulatory function in vitro and delay the SS disease activity in vivo. In conclusion, BMP6 could not only affect the secreting function of epithelial cells in the salivary gland but also influence the immunomodulatory properties of SGMSCs, which may trigger or enhance the autoimmune reflection in SS.
Collapse
Affiliation(s)
- Junji Xu
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China .,2 Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda, Maryland
| | - Yingying Su
- 3 Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University , Beijing, China
| | - Lei Hu
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Alexander Cain
- 2 Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda, Maryland
| | - Yi Gu
- 4 Department of Pediatrics, Beijing Chaoyang Hospital, Capital Medical University , Beijing, China
| | - Bowen Liu
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Ruiqing Wu
- 3 Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University , Beijing, China
| | - Songlin Wang
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Hao Wang
- 3 Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University , Beijing, China
| |
Collapse
|
120
|
Mesenchymal Stem Cells Alleviate DHEA-Induced Polycystic Ovary Syndrome (PCOS) by Inhibiting Inflammation in Mice. Stem Cells Int 2019; 2019:9782373. [PMID: 31611920 PMCID: PMC6757294 DOI: 10.1155/2019/9782373] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/12/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility in women of reproductive age. Chronic inflammation is considered to be the cause of ovarian dysfunction. Increasing evidence in animal studies and in preliminary clinical trials has demonstrated that MSCs possess immunomodulatory effects via their interaction with immune cells. However, their contribution to PCOS remains unclear. In this study, we showed that the administration of hUC-MSCs could efficiently improve the pathological changes of PCOS mice induced by dehydroepiandrosterone (DHEA), including ovarian histopathology and function. Moreover, we found that the administration of MSCs significantly downregulated the expression of proinflammatory factors (TNF-α, IL-1β, and IFN-γ) and fibrosis-related genes (CTGF) in ovarian and uterus tissues and affected the systemic inflammatory response. The percentage of peripheral neutrophils, M1 macrophages, and B cells was significantly reduced, while M2 macrophages and regulatory T cells (Tregs) were increased in hUC-MSC-treated mice. In the spleen, the percentage of neutrophils, M1 macrophages, IFN-γ+CD19+B cell, IFN-γ+CD4+T cells (Th1), and IL-17+CD4+T cells (Th17) was significantly decreased in hUC-MSC-treated mice. These results suggested that hUC-MSC treatment could alleviate ovarian dysfunction by inhibiting ovarian local and systemic inflammatory responses.
Collapse
|
121
|
Shi Y, Wang Y, Li Q, Liu K, Hou J, Shao C, Wang Y. Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases. Nat Rev Nephrol 2019; 14:493-507. [PMID: 29895977 DOI: 10.1038/s41581-018-0023-5] [Citation(s) in RCA: 706] [Impact Index Per Article: 141.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mesenchymal stem cells (MSCs; also referred to as mesenchymal stromal cells) have attracted much attention for their ability to regulate inflammatory processes. Their therapeutic potential is currently being investigated in various degenerative and inflammatory disorders such as Crohn's disease, graft-versus-host disease, diabetic nephropathy and organ fibrosis. The mechanisms by which MSCs exert their therapeutic effects are multifaceted, but in general, these cells are thought to enable damaged tissues to form a balanced inflammatory and regenerative microenvironment in the presence of vigorous inflammation. Studies over the past few years have demonstrated that when exposed to an inflammatory environment, MSCs can orchestrate local and systemic innate and adaptive immune responses through the release of various mediators, including immunosuppressive molecules, growth factors, exosomes, chemokines, complement components and various metabolites. Interestingly, even nonviable MSCs can exert beneficial effects, with apoptotic MSCs showing immunosuppressive functions in vivo. Because the immunomodulatory capabilities of MSCs are not constitutive but rather are licensed by inflammatory cytokines, the net outcomes of MSC activation might vary depending on the levels and the types of inflammation within the residing tissues. Here, we review current understanding of the immunomodulatory mechanisms of MSCs and the issues related to their therapeutic applications.
Collapse
Affiliation(s)
- Yufang Shi
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China. .,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Yu Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qing Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Keli Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianquan Hou
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, China
| | - Ying Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences/Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
122
|
Dehqan Niri A, Karimi Zarchi AA, Ghadiri Harati P, Salimi A, Mujokoro B. Tissue engineering scaffolds in the treatment of brain disorders in geriatric patients. Artif Organs 2019; 43:947-960. [DOI: 10.1111/aor.13485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/26/2019] [Accepted: 04/29/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Alireza Dehqan Niri
- Nanobiotechnology Research Center Baqiyatallah University of Medical Sciences Tehran Iran
| | | | - Parisa Ghadiri Harati
- Department of Physiotherapy, School of Rehabilitation Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Ali Salimi
- Nanobiotechnology Research Center Baqiyatallah University of Medical Sciences Tehran Iran
| | - Basil Mujokoro
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
123
|
Clearance of apoptotic cells by mesenchymal stem cells contributes to immunosuppression via PGE2. EBioMedicine 2019; 45:341-350. [PMID: 31248835 PMCID: PMC6642220 DOI: 10.1016/j.ebiom.2019.06.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022] Open
Abstract
Background Defective clearance of apoptotic cells (ACs) has been suggested to be involved in the pathogenesis of systemic lupus erythematosus (SLE). Mesenchymal stem cells (MSCs) exhibit promising therapeutic effects on SLE, but whether MSCs phagocytose ACs and contributes to the underlying mechanism in the treatment of SLE remain unknown. Methods Human umbilical cord (UC) MSCs were co-cultured with ACs, and the engulfment of ACs by MSCs was either detected by flow cytometry or observed under confocal laser scanning microscope. Peripheral blood mononuclear cells (PBMCs) from healthy controls (HCs) were cultured in MSC conditioned medium (MCM) or MSC exposed to ACs (AC-MSC) conditioned medium (ACMCM), and then CD4+ T cell proliferation was detected. Soluble factors including prostaglandin (PG)E2 in the supernatants of MSCs and AC-MSCs, as well as in the mouse peritoneal lavage fluids (PLF) were determined by enzyme-linked immunosorbent assay (ELISA). Cyclooxygenase (COX)2 inhibitors and siRNA transfection were utilized to determine the function of COX2/PGE2 in AC-MSC-mediated immunosuppression. PGE2 metabolites (PGEM) in the plasma of SLE patients were measured before and 24 h after MSC transplantation respectively. Findings Human UC MSCs possessed the ability to engulf ACs. AC-MSCs increased MSC-mediated suppression of CD4+ T cell proliferation compared to MSCs alone. Mechanistically, ACs stimulated MSCs to express COX2 and consequently produced PGE2 that inhibited T cell responses. NF-κB signalling pathway mediated the activation of COX2/PGE2 in AC-MSCs. Importantly, in patients with SLE, the plasma PGEM levels increased significantly in those with reduced apoptotic mononuclear cells in peripheral blood after MSC transplantation. Interpretation Clearance of ACs by MSCs contributes to immunosuppressive function via increasing PGE2 production. These findings reveal a previously unrecognized role of MSC-mediated phagocytosis of ACs in MSC-based immunotherapy. Fund This study was supported by grants from the Chinese Major International (Regional) Joint Research Project (No. 81720108020), the Jiangsu Province Major Research and Development Program (No. BE2015602) and the Jiangsu Province 333 Talent Grant (BRA2016001). WJ. Chen was supported by the Intramural Research Program of NIH, NIDCR.
Collapse
|
124
|
Kokorev OV, Khodorenko VN, Serebrov VY, Dambaev GT, Gunter VE. Co-Transplantation of Pancreatic Islet Cells and Mesenchymal Bone Marrow Precursors on Titanium Nickelide Scaffolds in Alloxan-Induced Diabetes Mellitus. Bull Exp Biol Med 2019; 167:140-144. [PMID: 31183647 DOI: 10.1007/s10517-019-04478-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Indexed: 01/04/2023]
Abstract
We studied therapeutic activity of co-transplantation of allogeneic pancreatic islet cells and mesenchymal bone marrow progenitors on TiNi scaffolds in Wistar rats with experimental alloxan-induced diabetes mellitus. In preliminary experiments with co-culturing of cells in different proportions followed by their transplantation on tissue-engineered constructs, the optimum ratio of these cells was determined - 3:1. Regeneration was assessed by biochemical methods by the blood levels of glucose and glycosylated hemoglobin on days 15, 30, and 5. In the group with combined cell transplantation on TiNi scaffold, normalization of the studied biochemical parameters occurred earlier than after monotherapy with allogenic islet cells and was associated with an increase in animal lifespan. Normalization of the parameters of bone marrow hemopoiesis, in particular, the number of myelokaryocytes and erythroblasts was also noted.
Collapse
Affiliation(s)
- O V Kokorev
- Research Institute of Medical Materials and Shape Memory Implants, National Research Tomsk State University, Tomsk, Russia. .,Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia.
| | - V N Khodorenko
- Research Institute of Medical Materials and Shape Memory Implants, National Research Tomsk State University, Tomsk, Russia
| | - V Yu Serebrov
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - G Ts Dambaev
- Siberian State Medical University, Ministry of Health of the Russian Federation, Tomsk, Russia
| | - V E Gunter
- Research Institute of Medical Materials and Shape Memory Implants, National Research Tomsk State University, Tomsk, Russia
| |
Collapse
|
125
|
The Immunomodulatory Potential of Wharton's Jelly Mesenchymal Stem/Stromal Cells. Stem Cells Int 2019; 2019:3548917. [PMID: 31281372 PMCID: PMC6594275 DOI: 10.1155/2019/3548917] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/01/2019] [Accepted: 05/22/2019] [Indexed: 12/13/2022] Open
Abstract
The benefits attributed to mesenchymal stem/stromal cells (MSC) in cell therapy applications are mainly attributed to the secretion of factors, which exhibit immunomodulatory and anti-inflammatory effects and stimulate angiogenesis. Despite the desirable features such as high proliferation levels, multipotency, and immune response regulation, there are important variables that must be considered. Although presenting similar morphological aspects, MSC collected from different tissues can form heterogeneous cellular populations and, therefore, manifest functional differences. Thus, the source of MSC should be a factor to be considered in the development of novel therapies. The following text presents an updated review of recent research outcomes related to Wharton's jelly mesenchymal stem/stromal cells (WJ-MSC), harvested from umbilical cords and considered novel and potential candidates for the development of cell-based approaches. This text highlights information on how WJ-MSC affect immune responses in comparison with other sources of MSC.
Collapse
|
126
|
Durcan L, O'Dwyer T, Petri M. Management strategies and future directions for systemic lupus erythematosus in adults. Lancet 2019; 393:2332-2343. [PMID: 31180030 DOI: 10.1016/s0140-6736(19)30237-5] [Citation(s) in RCA: 325] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2019] [Accepted: 01/23/2019] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterised by the loss of self-tolerance and formation of nuclear autoantigens and immune complexes resulting in inflammation of multiple organs. The clinical presentation of SLE is heterogeneous, can involve one or more organs, including the skin, kidneys, joints, and nervous system, and take a chronic or relapsing and remitting disease course. SLE is most common in women and in those of non-white ethnicity. Because of the multitude of presentations, manifestations, and serological abnormalities in patients with SLE, diagnosis can be challenging. Therapeutic approaches predominantly involve immunomodulation and immunosuppression and are targeted to the specific organ manifestation, with the aim of achieving low disease activity. Despite many treatment advances and improved diagnostics, SLE continues to cause substantial morbidity and premature mortality. Current management strategies, although helpful, are limited by high failure rates and toxicity. An overreliance on corticosteroid therapy contributes to much of the long-term organ damage. In this Seminar, we outline the classification criteria for SLE, current treatment strategies and medications, the evidence supporting their use, and explore potential future therapies.
Collapse
Affiliation(s)
- Laura Durcan
- Department of Rheumatology, Beaumont Hospital, Dublin, Ireland; Department of Medicine, The Royal College of Surgeons of Ireland, Dublin, Ireland.
| | - Tom O'Dwyer
- School of Physiotherapy, Trinity College, Dublin, Ireland
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MA, USA
| |
Collapse
|
127
|
Rota C, Morigi M, Imberti B. Stem Cell Therapies in Kidney Diseases: Progress and Challenges. Int J Mol Sci 2019; 20:ijms20112790. [PMID: 31181604 PMCID: PMC6600599 DOI: 10.3390/ijms20112790] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 05/31/2019] [Accepted: 06/05/2019] [Indexed: 12/15/2022] Open
Abstract
The prevalence of renal diseases is emerging as a public health problem. Despite major progress in supportive therapy, mortality rates among patients remain high. In an attempt to find innovative treatments to stimulate kidney regeneration, stem cell-based technology has been proposed as a potentially promising strategy. Here, we summarise the renoprotective potential of pluripotent and adult stem cell therapy in experimental models of acute and chronic kidney injury and we explore the different mechanisms at the basis of stem cell-induced kidney regeneration. Specifically, cell engraftment, incorporation into renal structures, or paracrine activities of embryonic or induced pluripotent stem cells as well as mesenchymal stem cells and renal precursors are analysed. We also discuss the relevance of stem cell secretome-derived bioproducts, including soluble factors and extracellular vesicles, and the option of using them as cell-free therapy to induce reparative processes. The translation of the experimental results into clinical trials is also addressed, highlighting the safety and feasibility of stem cell treatments in patients with kidney injury.
Collapse
Affiliation(s)
- Cinzia Rota
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy.
| | - Marina Morigi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy.
| | - Barbara Imberti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126 Bergamo, Italy.
| |
Collapse
|
128
|
Ye Z, Lu W, Liang L, Tang M, Wang Y, Li Z, Zeng H, Wang A, Lin M, Huang L, Wang H, Hu H. Mesenchymal stem cells overexpressing hepatocyte nuclear factor-4 alpha alleviate liver injury by modulating anti-inflammatory functions in mice. Stem Cell Res Ther 2019; 10:149. [PMID: 31133062 PMCID: PMC6537220 DOI: 10.1186/s13287-019-1260-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 05/03/2019] [Accepted: 05/10/2019] [Indexed: 01/20/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) can migrate to tissue injury sites where they can induce multipotential differentiation and anti-inflammation effects to treat tissue injury. When traditional therapeutic methods do not work, MSCs are considered to be one of the best candidates for cell therapy. MSCs have been used for treating several injury- and inflammation-associated diseases, including liver cirrhosis. However, the therapeutic effect of MSCs is limited. In some cases, the anti-inflammatory function of naïve MSCs is not enough to rescue tissue injury. Methods Carbon tetrachloride (CCl4) was used to establish a mouse liver cirrhosis model. Enhanced green fluorescence protein (EGFP) and hepatocyte nuclear factor-4α (HNF-4α) overexpression adenoviruses were used to modify MSCs. Three weeks after liver injury induction, mice were injected with bone marrow MSCs via their tail vein. The mice were then sacrificed 3 weeks after MSC injection. Liver injury was evaluated by measuring glutamic-pyruvic transaminase (ALT) and glutamic oxalacetic transaminase (AST) levels. Histological and molecular evaluations were performed to study the mechanisms. Results We found that HNF-4α-overexpressing MSCs had a better treatment effect than unmodified MSCs on liver cirrhosis. In the CCl4-induced mouse liver injury model, we found that HNF-4α-MSCs reduced inflammation in the liver and alleviated liver injury. In addition, we found that HNF-4α promoted the anti-inflammatory effect of MSCs by enhancing nitric oxide synthase (iNOS) expression, which was dependent on the nuclear factor kappa B (NF-κB) signalling pathway. Conclusions MSCs overexpressing HNF-4α exerted good therapeutic effects against mouse liver cirrhosis due to an enhanced anti-inflammatory effect. Gene modification is likely a promising method for improving the effects of cell therapy. Electronic supplementary material The online version of this article (10.1186/s13287-019-1260-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhenxiong Ye
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Wenfeng Lu
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China.,Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Lei Liang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China.,Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, China
| | - Min Tang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Yunfeng Wang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Zhen Li
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Heping Zeng
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Aili Wang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Moubin Lin
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Lei Huang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Hui Wang
- Department of General Surgery, Yangpu Hospital, Tongji University School of Medicine, 450 Teng Yue Road, Shanghai, 200090, China
| | - Hai Hu
- Center of Gallbladder Disease, Shanghai East Hospital, Tongji University School of Medicine, 150 Ji Mo Road, Shanghai, 201200, China.
| |
Collapse
|
129
|
Prognostic Factors for Clinical Response in Systemic Lupus Erythematosus Patients Treated by Allogeneic Mesenchymal Stem Cells. Stem Cells Int 2019; 2019:7061408. [PMID: 31191681 PMCID: PMC6525791 DOI: 10.1155/2019/7061408] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/22/2019] [Accepted: 02/12/2019] [Indexed: 12/22/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease with a broad range of clinical manifestations and a heterogeneous disease course. There is no cure for SLE, but current standard pharmacotherapies can improve disease prognosis in most patients. However, some patients are refractory to conventional treatments and require alternative treatment options. The present study is aimed at identifying predictors of clinical response to allogeneic bone marrow-derived or umbilical cord-derived mesenchymal stem cell (BM-/UC-MSC) transplant in SLE. All adult patients identified in the Nanjing database with an SLE Disease Activity Index (SLEDAI) score ≥ 8 at baseline that had undergone MSC transplant and who had at least 1 year of follow-up after one or two successive intravenous injections of allogeneic BM-/UC-MSCs (1 million/kg) were analyzed. SLE symptoms and SLEDAI were assessed at baseline and during follow-up to determine low disease activity (LDA) and clinical remission (CR) at 1, 3, 6, and 12 months. Sixty-nine patients were included in the study, with a median (range) SLEDAI of 13 (8-34) at baseline. Among the 69 patients, 40 (58%) achieved LDA and 16 (23%) achieved CR with a SLEDAI of 9 (4–20), 8 (0-16), 6 (0-18), and 5 (0-18) after 1, 3, 6, and 12 months, respectively. Older age (p = 0.006) and no arthralgia/arthritis at baseline (p = 0.03) were associated with a higher rate of LDA. Achieving CR was associated with older age (p = 0.033), no arthralgia/arthritis at baseline (p = 0.001), and no prior use of cyclophosphamide (p = 0.003) or hydroxychloroquine (p = 0.016). Future studies using unique immunosuppressive regimens and allogeneic MSC sources will further elucidate determinants of clinical response to MSC transplant in SLE.
Collapse
|
130
|
Wang J, Zhang Y, Cloud C, Duke T, Owczarski S, Mehrotra S, Adams DB, Morgan K, Gilkeson G, Wang H. Mesenchymal Stem Cells from Chronic Pancreatitis Patients Show Comparable Potency Compared to Cells from Healthy Donors. Stem Cells Transl Med 2019; 8:418-429. [PMID: 30680957 PMCID: PMC6477001 DOI: 10.1002/sctm.18-0093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are proven to be beneficial in islet transplantation, suggesting a potential therapeutic role of them in total pancreatectomy with islet autotransplantation (TP-IAT) for chronic pancreatitis (CP) patients. We investigated whether MSCs derived from CP patients are suitable for use in autologous cell therapy. MSCs from healthy donors (H-MSCs) and CP patients (CP-MSCs) were studied for phenotype, colony formation potential, multilineage differentiation ability, proliferation, senescence, secretory characters, and immunosuppressive functions. The potential protective effect of CP-MSCs was evaluated on hypoxia-induced islet cell death. Cell surface markers were similar between H-MSCs and CP-MSCs, as well as the ability of colony formation, multilineage differentiation, secretion of vascular endothelial growth factor and transforming growth factor (TGF-β), senescence, and inhibition of T cells proliferation in vitro. We found that growth differentiation factor 6 and hepatocyte growth factor (HGF) were significantly downregulated, whereas TGFβ and matrix metalloproteinase-2 were significantly upregulated in CP-MSCs compared with H-MSCs, among 84 MSC-related genes investigated in this study. MSCs from CP patients secreted less HGF, compared with the H-MSCs. A higher interferon-γ-induced indoleamine 2,3-dioxygenase expression was observed in CP-MSCs compared to H-MSCs. Moreover, CP-MSCs prevented hypoxia-induced β cell deaths to a similar extent as H-MSCs. Regardless of moderate difference in gene expression, CP-MSCs possess similar immunomodulatory and prosurvival functions to H-MSCs, and may be suitable for autologous cell therapy in CP patients undergoing TP-IAT. Stem Cells Translational Medicine 2019;8:418-429.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Yong Zhang
- College of Life ScienceQingdao Agricultural UniversityQingdaoPeople's Republic of China
| | - Colleen Cloud
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Tara Duke
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Stefanie Owczarski
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Shikhar Mehrotra
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - David B. Adams
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Katherine Morgan
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Gary Gilkeson
- Department of MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Hongjun Wang
- Department of SurgeryMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
131
|
Liu S, wang J, Han R, Meng M, Wang W, Zhao Y, Yang F, Yang L, Gao H, Zhao Y, Yang L, Wang R, Tang W, Li Y, Duan S, Wang J, He Z, Li L, Hou Z. Therapeutic effect of transplanted umbilical cord mesenchymal stem cells in a cynomolgus monkey model of multiple sclerosis. Am J Transl Res 2019; 11:2516-2531. [PMID: 31105859 PMCID: PMC6511768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/28/2019] [Indexed: 06/09/2023]
Abstract
Multiple sclerosis (MS) is a demyelinating disease affecting 2.5 million young people worldwide because of its immune-mediated pathological mechanisms. Recent studies have shown that stem cell transplantation is a new potential therapy for MS. There has been renewed interest in cell therapy to improve quality of life for MS patients. In this study, the experimental autoimmune encephalomyelitis (EAE) model, which is the most commonly model to mimic MS, was successfully established in cynomolgus monkeys. To evaluate the therapeutic effect of human umbilical cord mesenchymal stem cells (UCMSCs) on MS, we intravenously transplanted UCMSCs into cynomolgus monkeys with EAE. Our results showed that UCMSC transplantation significantly ameliorated the clinical symptoms of MS. Magnetic resonance imaging and clinical signs indicated that demyelination was obviously decreased after UCMSCs therapy. Moreover, the present study showed that the mechanisms, involved in the effects of UCMSCs on MS, included their immunomodulatory functions to regulate cytokine secretion and affect functional differentiation of the T cell lineage.
Collapse
Affiliation(s)
- Shijie Liu
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Jin wang
- Department of Radiology, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
| | - Rui Han
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Mingyao Meng
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Wenju Wang
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Yuan Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical CollegeKunming 650118, Yunnan Provience, P. R. China
| | - Fengmei Yang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical CollegeKunming 650118, Yunnan Provience, P. R. China
| | - Li Yang
- Department of Geriatrics, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
| | - Hui Gao
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Yiyi Zhao
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Lirong Yang
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Runqing Wang
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Weiwei Tang
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Yanyan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical CollegeKunming 650118, Yunnan Provience, P. R. China
| | - Suqin Duan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical CollegeKunming 650118, Yunnan Provience, P. R. China
| | - Junbing Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical CollegeKunming 650118, Yunnan Provience, P. R. China
| | - Zhanlong He
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical CollegeKunming 650118, Yunnan Provience, P. R. China
| | - Lin Li
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| | - Zongliu Hou
- Department of Central Laboratory, Yan’an Affiliated Hospital of Kunming Medical UniversityKunming 650051, Yunnan Provience, P. R. China
- Yunnan Cell Biology and Clinical Translation Research Center, Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan ProvinceKunming 650051, Yunnan Provience, P. R. China
| |
Collapse
|
132
|
Yan F, Liu O, Zhang H, Zhou Y, Zhou D, Zhou Z, He Y, Tang Z, Wang S. Human dental pulp stem cells regulate allogeneic NK cells' function via induction of anti-inflammatory purinergic signalling in activated NK cells. Cell Prolif 2019; 52:e12595. [PMID: 30953394 PMCID: PMC6536423 DOI: 10.1111/cpr.12595] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/17/2019] [Accepted: 01/30/2019] [Indexed: 12/20/2022] Open
Abstract
Objectives Mesenchymal stem cells (MSCs) could regulate the function of various immune cells. It remains unclear whether MSCs additionally possess immunostimulatory properties. We investigated the impact of human MSCs on the responsiveness of primary natural killer (NK) cells in terms of induction of anti‐inflammatory purinergic signalling. Material and Methods We obtained human bone marrow mesenchymal stem cells (BMMSCs) and dental pulp stem cells (DPSCs). NK cells were isolated from peripheral blood of healthy volunteers. Activated NK cells were cultured with MSCs. Proliferation assay, apoptosis analysis, activating or inhibitory receptor expression and degranulation assay were used to explore NK cells’ function. High‐performance liquid chromatography was used to investigate the purinergic signalling in activated NK cells. Results Both DPSCs and BMMSCs could impair proliferation and promote apoptosis of activated NK cells. Also, activated NK cells could cause DPSCs to lyse. Furthermore, the expression of activating NK cells’ receptors was decreased, but inhibitory receptors of NK cells were elevated following co‐cultivation. NK cells acquired CD73 expression, while MSCs could release ATP into the extracellular space where nucleotides were converted into adenosine (ADO) following co‐culture system. Under the existence of exogenous 2‐chloroadenosine (CADO), the cytotoxic capacity of NK cells was remarkably depressed in a concentration‐dependent manner. Conclusions DPSCs and BMMSCs could depress NK cells’ function by hydrolysing ATP to ADO using CD39 and CD73 enzymatic activity. Our data suggested that DPSCs might represent a new strategy for treating immune‐related diseases by regulating previously unrecognized functions in innate immune responses.
Collapse
Affiliation(s)
- Fei Yan
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China.,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Ousheng Liu
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China.,Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Haixia Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yueying Zhou
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Dian Zhou
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zekun Zhou
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Yuhong He
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Zhangui Tang
- Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, Hunan, China
| | - Songlin Wang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
133
|
Vasilev G, Ivanova M, Ivanova-Todorova E, Tumangelova-Yuzeir K, Krasimirova E, Stoilov R, Kyurkchiev D. Secretory factors produced by adipose mesenchymal stem cells downregulate Th17 and increase Treg cells in peripheral blood mononuclear cells from rheumatoid arthritis patients. Rheumatol Int 2019; 39:819-826. [PMID: 30944956 DOI: 10.1007/s00296-019-04296-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023]
Abstract
We aimed to assess the immunoregulatory effects of secretory factors produced by adipose tissue-derived MSC (AT-MSC) on Th17 and Treg subsets from patients with rheumatoid arthritis (RA). 17 patients with active disease matching the ACR/EULAR 2010 criteria for RA were included. Patients' peripheral blood mononuclear cells (PBMC) were cultured in AT-MSC-conditioned medium (AT-MSCcm) and in control medium. The cytokine production of AT-MSC and PBMC was quantified by ELISA. Th17 and Treg were determined by flow cytometry. AT-MSCcm contained: IL-6, IL-17, IL-21, CCL2, CCL5, IL-8, sVEGF-A and PGE2. Cultivation of patients' PBMC with AT-MSCcm increased TGF-β1 (8318 pg/ml; IQR 6327-11,686) vs control medium [6227 pg/ml (IQR 1681-10,148, p = 0.013)]. PBMC cultivated with AT-MSCcm downregulated TNF-α, IL-17A, and IL-21 compared to control PBMC: 5 pg/ml IQR (1.75-11.65) vs 1 pg/ml (IQR 0.7-1.9), p = 0.001; 4.2 pg/ml (IQR 3.1-6.1) vs 2.3 pg/ml (IQR.75-5.42), p = 0.017; 66.9 pg/ml (IQR 40.6-107.2) vs 53 pg/ml (IQR 22-73), p = 0.022. Th17 decreased under the influence of AT-MSCcm: 10.13 ± 3.88% vs 8.98 ± 3.58%, p = 0.02. CD4+FoxP3+, CD4+CD25-FoxP3+, and CD4+CD25+FoxP3+ was 11.35 ± 4.1%; 7.13 ± 3.12% and 4.22 ± 2% in control PBMC. Accordingly, CD4+FoxP3+, CD4+CD25-FoxP3+, and CD4+CD25+FoxP3+ significantly increased in PBMC cultured with AT-MSCcm: 15.6 ± 6.1%, p = 0.001; 9.56 ± 5.4%, p = 0.004 and 6.04 ± 3.6%, p = 0.001. All these effects could define MSC-based approaches as adequate avenues for further treatment development in RA.
Collapse
Affiliation(s)
- Georgi Vasilev
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria.
| | - Mariana Ivanova
- Clinic of Rheumatology, University Hospital "St. Ivan Rilski"-Sofia, Department of Internal Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Ekaterina Ivanova-Todorova
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| | - Kalina Tumangelova-Yuzeir
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| | - Ekaterina Krasimirova
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| | - Rumen Stoilov
- Clinic of Rheumatology, University Hospital "St. Ivan Rilski"-Sofia, Department of Internal Medicine, Medical University of Sofia, Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, Department of Clinical Immunology, University Hospital "St. Ivan Rilski"-Sofia, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
134
|
Tang X, Li W, Wen X, Zhang Z, Chen W, Yao G, Chen H, Wang D, Shi S, Sun L. Transplantation of dental tissue-derived mesenchymal stem cells ameliorates nephritis in lupus mice. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:132. [PMID: 31157253 DOI: 10.21037/atm.2019.02.41] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Recently, clinical studies have suggested that transplantation of umbilical cord mesenchymal stem cells (UC-MSCs) were able to alleviate clinical symptoms of refractory systemic lupus erythematosus (SLE). Although dental tissue derived MSCs, including dental pulp stem cells (DPSCs) and periodontal ligament stem cell (PDLSCs), have been reported to possess immunomodulatory functions, whether they can ameliorate SLE symptoms as UC-MSCs remains to be elucidated. Methods We assessed the abilities of DPSCs and PDLSCs to treat SLE, cells were transferred intravenously to 28-week old B6/lpr mice. Ten weeks later, mice were sacrificed. Serum anti-dsDNA antibodies and anti-nuclear antibodies (ANA) were measured by ELISA. Renal pathology was analyzed by H&E, PAS and MASSON staining. Aggregation of IgG and IgM in the glomerulus was examined by immunofluorescence. Frequencies of Th1, Th2, Treg, Th17, Tfh, and plasma cells were determined by surface and intracellular staining. Serum IL-6, IL-10, IL-17 and MCP-1 were measured by Milliplex® MAP technology. Results Same as UC-MSCs, both DPSCs and PDLSCs could efficiently downregulate 24-h proteinuria, anti-dsDNA antibodies and glomerular IgG/IgM in B6/lpr mice. However, DPSCs but not PDLSCs could ameliorate the glomerular lesion in B6/lpr mice. Compared to the phosphate buffered saline (PBS) group, percentages of Th1 (CD4+IFNγ+) cells and plasma (B220-CD138+) cells in the spleen were significantly decreased in DPSCs and PDLSCs groups. There was no significant difference in Th2 (CD4+IL4+), Th17 (CD4+IL17+), Tfh (CD4+PD-1+CXCR5+) and Treg (CD4+CD25+Foxp3+) cells. Serum IL-6, IL-10, IL-17 and MCP-1 levels didn't change after MSCs transplantation. Conclusions Our results show that both DPSCs and PDLSCs can alleviate the disease symptoms of lupus-prone B6/lpr mice. DPSCs are also effective in reducing kidney glomerular lesion and perivascular inflammation infiltration as well as UC-MSCs, suggesting that DPSCs might be another choice for SLE treatment.
Collapse
Affiliation(s)
- Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wenchao Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xin Wen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zhuoya Zhang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Hongwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Songtao Shi
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
135
|
Geng L, Tang X, Zhou K, Wang D, Wang S, Yao G, Chen W, Gao X, Chen W, Shi S, Shen N, Feng X, Sun L. MicroRNA-663 induces immune dysregulation by inhibiting TGF-β1 production in bone marrow-derived mesenchymal stem cells in patients with systemic lupus erythematosus. Cell Mol Immunol 2019; 16:260-274. [PMID: 30886422 PMCID: PMC6460486 DOI: 10.1038/cmi.2018.1] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 12/17/2017] [Accepted: 12/22/2017] [Indexed: 02/08/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are critical for immune regulation. Although several microRNAs (miRNAs) have been shown to participate in autoimmune pathogenesis by affecting lymphocyte development and function, the roles of miRNAs in MSC dysfunction in autoimmune diseases remain unclear. Here, we show that patients with systemic lupus erythematosus (SLE) display a unique miRNA signature in bone marrow-derived MSCs (BMSCs) compared with normal controls, among which miR-663 is closely associated with SLE disease activity. MiR-663 inhibits the proliferation and migration of BMSCs and impairs BMSC-mediated downregulation of follicular T helper (Tfh) cells and upregulation of regulatory T (Treg) cells by targeting transforming growth factor β1 (TGF-β1). MiR-663 overexpression weakens the therapeutic effect of BMSCs, while miR-663 inhibition improves the remission of lupus disease in MRL/lpr mice. Thus, miR-663 is a key mediator of SLE BMSC regulation and may serve as a new therapeutic target for the treatment of lupus.
Collapse
Affiliation(s)
- Linyu Geng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Kangxing Zhou
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Shiying Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China
| | - Xiang Gao
- Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, 210000, Nanjing, China
| | - Wanjun Chen
- Mucosal Immunology Section, OPCB, National Institute of Dental and Craniofacial Research, National Institutes of Health, 20892-2190, Bethesda, MD, USA
| | - Songtao Shi
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, 19104-6004, Philadelphia, PA, USA
| | - Nan Shen
- Joint Molecular Rheumatology Laboratory of the Institute of Health Sciences and Shanghai Renji Hospital, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, and Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuebing Feng
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, 210008, Nanjing, China.
| |
Collapse
|
136
|
Miranda JP, Camões SP, Gaspar MM, Rodrigues JS, Carvalheiro M, Bárcia RN, Cruz P, Cruz H, Simões S, Santos JM. The Secretome Derived From 3D-Cultured Umbilical Cord Tissue MSCs Counteracts Manifestations Typifying Rheumatoid Arthritis. Front Immunol 2019; 10:18. [PMID: 30804924 PMCID: PMC6370626 DOI: 10.3389/fimmu.2019.00018] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/04/2019] [Indexed: 01/23/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder whose treatment is mostly restricted to pain and symptom management and to the delay of joint destruction. Mesenchymal stem/stromal cells from the umbilical cord tissue (UC-MSCs) have previously been proven to be immunomodulatory and more efficient than bone marrow-derived MSCs in causing remission of local and systemic arthritic manifestations in vivo. Given the paracrine nature of UC-MSC activity, their application as active substances can be replaced by their secretome, thus avoiding allogeneic rejection and safety issues related to unwanted grafting. In this work, we aimed at demonstrating the viability of applying the 3D-primed UC-MSC secretome for the amelioration of arthritic signs. A proteomic analysis was performed to both, media conditioned by UC-MSC monolayer (CM2D) and 3D cultures (CM3D). The analysis of relevant trophic factors confirmed secretome profiles with very significant differences in terms of therapeutic potential. Whereas, CM3D was characterised by a prevailing expression of anti-inflammatory cytokines such as IL-10 and LIF, along with trophic factors involved in different mechanisms leading to tissue regeneration, such as PDGF-BB, FGF-2, I-309, SCF, and GM-CSF; CM2D presented relatively higher levels of IL-6, MCP-1, and IL-21, with recognised pro-inflammatory roles in joint disease and pleiotropic effects in the progression of rheumatoid arthritis (RA). Accordingly, different motogenic effects over mouse chondrocytes and distinct capacities of inducing glycosaminoglycan synthesis in vitro were observed between CM3D and CM2D. Finally, the evaluation of arthritic manifestations in vivo, using an adjuvant-induced model for arthritis (AIA), suggested a significantly higher therapeutic potential of CM3D over CM2D and even UC-MSCs. Histological analysis confirmed a faster remission of local and systemic arthritic manifestations of CM3D-treated animals. Overall, the results show that the use of UC-MSC CM3D is a viable and better strategy than direct UC-MSC administration for counteracting AIA-related signs. This strategy represents a novel MSC-based but nonetheless cell-free treatment for arthritic conditions such as those characterising RA.
Collapse
Affiliation(s)
- Joana P Miranda
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Sérgio P Camões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Maria M Gaspar
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Joana S Rodrigues
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Manuela Carvalheiro
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | | | | | | | - Sandra Simões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Jorge M Santos
- ECBio S.A., Amadora, Portugal.,Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente, Universidade do Porto, Porto, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
137
|
Myneni VD, McClain-Caldwell I, Martin D, Vitale-Cross L, Marko K, Firriolo JM, Labow BI, Mezey E. Mesenchymal stromal cells from infants with simple polydactyly modulate immune responses more efficiently than adult mesenchymal stromal cells. Cytotherapy 2018; 21:148-161. [PMID: 30595353 DOI: 10.1016/j.jcyt.2018.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 11/04/2018] [Accepted: 11/29/2018] [Indexed: 12/23/2022]
Abstract
Bone marrow-derived stromal cells or mesenchymal stromal cells (BMSCs or MSCs, as we will call them in this work) are multipotent progenitor cells that can differentiate into osteoblasts, adipocytes and chondrocytes. In addition, MSCs have been shown to modulate the function of a variety of immune cells. Donor age has been shown to affect the regenerative potential, differentiation, proliferation and anti-inflammatory potency of MSCs; however, the impact of donor age on their immunosuppressive activity is unknown. In this study, we evaluated the ability of MSCs derived from very young children and adults on T-cell suppression and cytokine secretion by monocytes/macrophages. MSCs were obtained from extra digits of children between 10 and 21 months and adults between 28 and 64 years of age. We studied cell surface marker expression, doubling time, lineage differentiation potential and immunosuppressive function of the MSCs. Young MSCs double more quickly and differentiate into bone and fat cells more efficiently than those from older donors. They also form more and dense colonies of fibroblasts (colony forming unit-fibroblast [CFU-F]). MSCs from both young and adult subjects suppressed T-cell proliferation in a mitogen-induced assay at 1:3 and 1:30 ratios. At a 1:30 ratio, however, MSCs from adults did not, but MSCs from infants did suppress T-cell proliferation. In the mixed lymphocyte reaction assay, MSCs from infants produced similar levels of suppression at all three MSC/T-cell ratios, but adult MSCs only inhibited T-cell proliferation at a 1:3 ratio. Cytokine analyses of co-cultures of MSCs and macrophages showed that both adult and young MSCs suppress tumor necrosis factor alpha (TNF-α) and induce interleukin-10 (IL-10) production in macrophage co-culture assay in a similar manner. Overall, this work shows that developing MSCs display a higher level of immunosuppression than mature MSCs.
Collapse
Affiliation(s)
- Vamsee D Myneni
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Ian McClain-Caldwell
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Martin
- Genomics & Computational Biology Core, National Institute of Dental and Craniofacial Research, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, Maryland, USA
| | - Lynn Vitale-Cross
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Karoly Marko
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph M Firriolo
- Department of Plastic and Oral Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Brian I Labow
- Department of Plastic and Oral Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eva Mezey
- Adult Stem Cell Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
138
|
Rufaihah AJ, Cheyyatraivendran S, Mazlan MDM, Lim K, Chong MSK, Mattar CNZ, Chan JKY, Kofidis T, Seliktar D. The Effect of Scaffold Modulus on the Morphology and Remodeling of Fetal Mesenchymal Stem Cells. Front Physiol 2018; 9:1555. [PMID: 30622472 PMCID: PMC6308149 DOI: 10.3389/fphys.2018.01555] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/17/2018] [Indexed: 12/17/2022] Open
Abstract
Hydrogel materials have been successfully used as matrices to explore the role of biophysical and biochemical stimuli in directing stem cell behavior. Here, we present our findings on the role of modulus in guiding bone marrow fetal mesenchymal stem cell (BMfMSC) fate determination using semi-synthetic hydrogels made from PEG-fibrinogen (PF). The BMfMSCs were cultivated in the PF for up to 2 weeks to study the influence of matrix modulus (i.e., cross-linking density of the PF) on BMfMSC survival, morphology and integrin expression. Both two-dimensional (2D) and three-dimensional (3D) culture conditions were employed to examine the BMfMSCs as single cells or as cell spheroids. The hydrogel modulus affected the rate of BMfMSC metabolic activity, the integrin expression levels and the cell morphology, both as single cells and as spheroids. The cell seeding density was also found to be an important parameter of the system in that high densities were favorable in facilitating more cell-to-cell contacts that favored higher metabolic activity. Our findings provide important insight about design of a hydrogel scaffold that can be used to optimize the biological response of BMfMSCs for various tissue engineering applications.
Collapse
Affiliation(s)
- Abdul Jalil Rufaihah
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suganya Cheyyatraivendran
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Muhammad Danial Mohd Mazlan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kenrich Lim
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mark Seow Khoon Chong
- Division of Bioengineering, School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | | | - Jerry Kok Yen Chan
- Department of Obstretics and Gynaecology, National University of Singapore, Singapore, Singapore
| | - Theodoros Kofidis
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Centre Singapore, National University Health System, Singapore, Singapore
| | - Dror Seliktar
- Nanoscience and Nanotechnology Initiative, National University of Singapore, Singapore, Singapore.,Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
139
|
Ferreira JR, Teixeira GQ, Santos SG, Barbosa MA, Almeida-Porada G, Gonçalves RM. Mesenchymal Stromal Cell Secretome: Influencing Therapeutic Potential by Cellular Pre-conditioning. Front Immunol 2018; 9:2837. [PMID: 30564236 PMCID: PMC6288292 DOI: 10.3389/fimmu.2018.02837] [Citation(s) in RCA: 333] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 11/16/2018] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are self-renewing, culture-expandable adult stem cells that have been isolated from a variety of tissues, and possess multipotent differentiation capacity, immunomodulatory properties, and are relatively non-immunogenic. Due to this unique set of characteristics, these cells have attracted great interest in the field of regenerative medicine and have been shown to possess pronounced therapeutic potential in many different pathologies. MSCs' mode of action involves a strong paracrine component resulting from the high levels of bioactive molecules they secrete in response to the local microenvironment. For this reason, MSCs' secretome is currently being explored in several clinical contexts, either using MSC-conditioned media (CM) or purified MSC-derived extracellular vesicles (EVs) to modulate tissue response to a wide array of injuries. Rather than being a constant mixture of molecular factors, MSCs' secretome is known to be dependent on the diverse stimuli present in the microenvironment that MSCs encounter. As such, the composition of the MSCs' secretome can be modulated by preconditioning the MSCs during in vitro culture. This manuscript reviews the existent literature on how preconditioning of MSCs affects the therapeutic potential of their secretome, focusing on MSCs' immunomodulatory and regenerative features, thereby providing new insights for the therapeutic use of MSCs' secretome.
Collapse
Affiliation(s)
- Joana R Ferreira
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Graciosa Q Teixeira
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Susana G Santos
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Mário A Barbosa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| | - Raquel M Gonçalves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
140
|
Li J, Xu SQ, Zhao YM, Yu S, Ge LH, Xu BH. Comparison of the biological characteristics of human mesenchymal stem cells derived from exfoliated deciduous teeth, bone marrow, gingival tissue, and umbilical cord. Mol Med Rep 2018; 18:4969-4977. [PMID: 30272340 PMCID: PMC6236220 DOI: 10.3892/mmr.2018.9501] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 08/30/2018] [Indexed: 12/24/2022] Open
Abstract
Different sources of mesenchymal stem cells (MSCs) may differ in their biological characteristics, which are important for their clinical application. In the present study, MSCs were isolated from human exfoliated deciduous teeth (SHED), bone marrow, gingival tissue and umbilical cord tissue, and their biological characteristics including surface markers, proliferation capacity, tumorigenicity and immunogenicity were analyzed by flow cytometric analysis, ELISA and co‑culture with human lymphocytes, respectively. The results indicated that all four types of stem cells obtained from different sources expressed MSC surface markers, and they did not show tumorigenicity either in vivo or in vitro. Stem cells from SHED exhibited the strongest proliferation capacity. Umbilical cord‑derived MSCs displayed the strongest immunomodulatory ability, while bone marrow MSCs exhibited the best antigen‑presenting potential in response to interferon‑γ stimulation. These results provide information on MSCs derived from different tissues, which may be helpful in their clinical application.
Collapse
Affiliation(s)
- Jing Li
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Shi-Qing Xu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Yu-Ming Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Shi Yu
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China
| | - Li-Hong Ge
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing 100081, P.R. China,Correspondence to: Dr Li-Hong Ge, Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, 22 Zhongguancun Avenue South, Haidian, Beijing 100081, P.R. China, E-mail:
| | - Bao-Hua Xu
- Dental Medical Center, China-Japan Friendship Hospital, Beijing 100029, P.R. China,Dr Bao-Hua Xu, Dental Medical Center, China-Japan Friendship Hospital, 2 East Cherry Street, Chaoyang, Beijing 100029, P.R. China, E-mail:
| |
Collapse
|
141
|
Mosanya CH, Isaacs JD. Tolerising cellular therapies: what is their promise for autoimmune disease? Ann Rheum Dis 2018; 78:297-310. [PMID: 30389690 PMCID: PMC6390030 DOI: 10.1136/annrheumdis-2018-214024] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 09/22/2018] [Accepted: 10/06/2018] [Indexed: 12/11/2022]
Abstract
The current management of autoimmunity involves the administration of immunosuppressive drugs coupled to symptomatic and functional interventions such as anti-inflammatory therapies and hormone replacement. Given the chronic nature of autoimmunity, however, the ideal therapeutic strategy would be to reinduce self-tolerance before significant tissue damage has accrued. Defects in, or defective regulation of, key immune cells such as regulatory T cells have been documented in several types of human autoimmunity. Consequently, it has been suggested that the administration of ex vivo generated, tolerogenic immune cell populations could provide a tractable therapeutic strategy. Several potentially tolerogenic cellular therapies have been developed in recent years; concurrent advances in cell manufacturing technologies promise scalable, affordable interventions if safety and efficacy can be demonstrated. These therapies include mesenchymal stromal cells, tolerogenic dendritic cells and regulatory T cells. Each has advantages and disadvantages, particularly in terms of the requirement for a bespoke versus an ‘off-the-shelf’ treatment but also their suitability in particular clinical scenarios. In this review, we examine the current evidence for these three types of cellular therapy, in the context of a broader discussion around potential development pathway(s) and their likely future role. A brief overview of preclinical data is followed by a comprehensive discussion of human data.
Collapse
Affiliation(s)
- Chijioke H Mosanya
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - John D Isaacs
- Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK .,Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
142
|
Azithromycin Promotes the Osteogenic Differentiation of Human Periodontal Ligament Stem Cells after Stimulation with TNF- α. Stem Cells Int 2018; 2018:7961962. [PMID: 30515223 PMCID: PMC6234456 DOI: 10.1155/2018/7961962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/18/2018] [Accepted: 06/21/2018] [Indexed: 12/19/2022] Open
Abstract
Background and Objective This study investigated the effects and underlying mechanisms of azithromycin (AZM) treatment on the osteogenic differentiation of human periodontal ligament stem cells (PDLSCs) after their stimulation with TNF-α in vitro. Methods. PDLSCs were isolated from periodontal ligaments from extracted teeth, and MTS assay was used to evaluate whether AZM and TNF-α had toxic effects on PDLSCs viability and proliferation. After stimulating PDLSCs with TNF-α and AZM, we analyzed alkaline phosphatase staining, alkaline phosphatase activity, and alizarin red staining to detect osteogenic differentiation. Real-time quantitative polymerase chain reaction (RT-qPCR) analysis was performed to detect the mRNA expression of osteogenic-related genes, including RUNX2, OCN, and BSP. Western blotting was used to measure the NF-κB signaling pathway proteins p65, phosphorylated p65, IκB-α, phosphorylated IκB-α, and β-catenin as well as the apoptosis-related proteins caspase-8 and caspase-3. Annexin V assay was used to detect PDLSCs apoptosis. Results TNF-α stimulation of PDLSCs decreased alkaline phosphatase and alizarin red staining, alkaline phosphatase activity, and mRNA expression of RUNX2, OCN, and BSP in osteogenic-conditioned medium. AZM enhanced the osteogenic differentiation of PDLSCs that were stimulated with TNF-α. Western blot analysis showed that β-catenin, phosphorated p65, and phosphorylated IκB-α protein expression decreased in PDLSCs treated with AZM. In addition, pretreatment of PDLSCs with AZM (10 μg/ml, 20 μg/ml) prevented TNF-α-induced apoptosis by decreasing caspase-8 and caspase-3 expression. Conclusions Our results showed that AZM promotes PDLSCs osteogenic differentiation in an inflammatory microenvironment by inhibiting the WNT and NF-κB signaling pathways and by suppressing TNF-α-induced apoptosis. This suggests that AZM has potential as a clinical therapeutic for periodontitis.
Collapse
|
143
|
Mardpour S, Hamidieh AA, Taleahmad S, Sharifzad F, Taghikhani A, Baharvand H. Interaction between mesenchymal stromal cell-derived extracellular vesicles and immune cells by distinct protein content. J Cell Physiol 2018; 234:8249-8258. [PMID: 30378105 DOI: 10.1002/jcp.27669] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Accepted: 10/02/2018] [Indexed: 12/13/2022]
Abstract
Mesenchymal stromal cells (MSCs) can effectively contribute to tissue regeneration inside the inflammatory microenvironment mostly through modulating immune responses. MSC-derived extracellular vesicles (MSC-EVs) display immunoregulatory functions similar to parent cells. Interactions between MSC-EVs and immune cells make them an ideal therapeutic candidate for infectious, inflammatory, and autoimmune diseases. These properties of MSC-EVs have encouraged researchers to perform extensive studies on multiple factors that mediate MSC-EVs immunomodulatory effects. Investigation of proteins involved in the complex interplay of MSC-EVs and immune cells may help us to better understand their functions. Here, we performed a comprehensive proteomic analysis of MSC-EVs that was previously reported by ExoCarta database. A total of 938 proteins were identified as MSC-EV proteome using quantitative proteomics techniques. Kyoto Encyclopedia of Genes and Genomes analysis demonstrates that ECM-receptor interaction, focal adhesion, and disease-specific pathways are enriched in MSC-EVs. By detail analysis of proteins presence in immune system process, we found that expression of some cytokines, chemokines, and chemokine receptors such as IL10, HGF, LIF, CCL2, VEGFC, and CCL20, which leads to migration of MSC-EVs to injured sites, suppression of inflammation and promotion of regeneration in inflammatory and autoimmune diseases. Also, some chemoattractant proteins such as CXCL2, CXCL8, CXCL16, DEFA1, HERC5, and IFITM2 were found in MSC-EV proteome. They may actively recruit immune cells to the proximity of MSC or MSC-EVs, may result in boosting immune response under specific circumstances, and may have protective role in infectious diseases. In this review, we summarize available information about immunomodulation of MSC-EVs with particular emphasis on their proteomics analysis.
Collapse
Affiliation(s)
- Soura Mardpour
- Tissue Engineering and Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Taleahmad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Farzaneh Sharifzad
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Adeleh Taghikhani
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
144
|
Gugjoo MB, Amarpal, Makhdoomi DM, Sharma GT. Equine Mesenchymal Stem Cells: Properties, Sources, Characterization, and Potential Therapeutic Applications. J Equine Vet Sci 2018; 72:16-27. [PMID: 30929778 DOI: 10.1016/j.jevs.2018.10.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 09/06/2018] [Accepted: 10/05/2018] [Indexed: 02/07/2023]
Abstract
Properties like sustained multiplication and self-renewal, and homing and multilineage differentiation to undertake repair of the damaged tissues make stem cells the lifeline for any living system. Therefore, stem cell therapy is regarded to carry immense therapeutic potential. Though the dearth of understanding about the basic biological properties and pathways involved in therapeutic benefits currently limit the application of stem cells in humans as well as animals, there are innumerable reports that suggest clinical benefits of stem cell therapy in equine. Among various stem cell sources, currently adult mesenchymal stem cells (MSCs) are preferred for therapeutic application in horse owing to their easy availability, capacity to modulate inflammation, and promote healing. Also the cells carry very limited teratogenic risk compared to the pluripotent stem cells. Mesenchymal stem cells were earlier considered mainly for musculoskeletal tissues, but now may also be utilized in other diverse clinical problems in horse, and the results may be extrapolated even for human medicine. The current review highlights biological properties, sources, mechanisms, and potential therapeutic applications of stem cells in equine practice.
Collapse
Affiliation(s)
- Mudasir Bashir Gugjoo
- Division of Surgery, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India.
| | - Amarpal
- Division of Surgery, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India
| | - Dil Mohammad Makhdoomi
- Division of Surgery, Faculty of Veterinary Sciences and Animal Husbandry, Shuhama, SKUAST-Kashmir, Srinagar, J&K, India
| | - Gutulla Taru Sharma
- Division of Physiology and Climatology, Indian Veterinary Research Institute-Izatnagar, Bareilly, UP, India
| |
Collapse
|
145
|
Liu D, Kou X, Chen C, Liu S, Liu Y, Yu W, Yu T, Yang R, Wang R, Zhou Y, Shi S. Circulating apoptotic bodies maintain mesenchymal stem cell homeostasis and ameliorate osteopenia via transferring multiple cellular factors. Cell Res 2018; 28:918-933. [PMID: 30030518 PMCID: PMC6123409 DOI: 10.1038/s41422-018-0070-2] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 04/26/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022] Open
Abstract
In the human body, 50-70 billion cells die every day, resulting in the generation of a large number of apoptotic bodies. However, the detailed biological role of apoptotic bodies in regulating tissue homeostasis remains unclear. In this study, we used Fas-deficient MRL/lpr and Caspase 3-/- mice to show that reduction of apoptotic body formation significantly impaired the self-renewal and osteo-/adipo-genic differentiation of bone marrow mesenchymal stem cells (MSCs). Systemic infusion of exogenous apoptotic bodies rescued the MSC impairment and also ameliorated the osteopenia phenotype in MRL/lpr, Caspase 3-/- and ovariectomized (OVX) mice. Mechanistically, we showed that MSCs were able to engulf apoptotic bodies via integrin αvβ3 and reuse apoptotic body-derived ubiquitin ligase RNF146 and miR-328-3p to inhibit Axin1 and thereby activate the Wnt/β-catenin pathway. Moreover, we used a parabiosis mouse model to reveal that apoptotic bodies participated in the circulation to regulate distant MSCs. This study identifies a previously unknown role of apoptotic bodies in maintaining MSC and bone homeostasis in both physiological and pathological contexts and implies the potential use of apoptotic bodies to treat osteoporosis.
Collapse
Affiliation(s)
- Dawei Liu
- Department of Orthodontics, Peking University School & Hospital of Stomatology, #22 Zhongguancun South Avenue, Beijing, 100081, China
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA
| | - Xiaoxing Kou
- Department of Orthodontics, Peking University School & Hospital of Stomatology, #22 Zhongguancun South Avenue, Beijing, 100081, China
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Chider Chen
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Shiyu Liu
- School of Stomatology, Fourth Military Medical University, Xi'an, Shanxi, 710032, China
| | - Yao Liu
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Wenjing Yu
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Tingting Yu
- Department of Orthodontics, Peking University School & Hospital of Stomatology, #22 Zhongguancun South Avenue, Beijing, 100081, China
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Ruili Yang
- Department of Orthodontics, Peking University School & Hospital of Stomatology, #22 Zhongguancun South Avenue, Beijing, 100081, China
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Runci Wang
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA
| | - Yanheng Zhou
- Department of Orthodontics, Peking University School & Hospital of Stomatology, #22 Zhongguancun South Avenue, Beijing, 100081, China
| | - Songtao Shi
- Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, PA, 19104, USA.
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA, 90033, USA.
| |
Collapse
|
146
|
Wang Z, Sun D. Adipose-Derived Mesenchymal Stem Cells: A New Tool for the Treatment of Renal Fibrosis. Stem Cells Dev 2018; 27:1406-1411. [PMID: 30032706 DOI: 10.1089/scd.2017.0304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As chronic kidney disease progresses, kidney tissue inevitably undergoes cell loss, accumulation of extracellular matrix, and kidney tissue fibrosis, eventually leading to end-stage renal disease. With the continuous innovation of cell therapy technology, mesenchymal stem cells are used in numerous fields, including cardiovascular diseases, diabetes, and kidney tissue injury repair. Adipose-derived mesenchymal stem cells (AMSCs), a type of pluripotent stem cells, have the potential for self-renewal and proliferation with low immunogenicity and significant anti-inflammatory properties. AMSCs can promote impaired cell regeneration and remodeling in renal lesions, thus avoiding further worsening of renal disease and even blocking or reversing the process of renal fibrosis. In this review, we discuss the mechanisms involved in the treatment of renal fibrosis with AMSCs and summarize the potential hazards that may exist in cell therapy.
Collapse
Affiliation(s)
- Zhuojun Wang
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, China
| | - Dong Sun
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, China .,2 Department of Internal Medicine and Diagnostics, Xuzhou Medical University , Xuzhou, China
| |
Collapse
|
147
|
Maxillary Sinus Lift Using Autologous Periosteal Micrografts: A New Regenerative Approach and a Case Report of a 3-Year Follow-Up. Case Rep Dent 2018; 2018:3023096. [PMID: 30140472 PMCID: PMC6081519 DOI: 10.1155/2018/3023096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/14/2018] [Indexed: 12/30/2022] Open
Abstract
This case report discusses about an innovative bone regeneration method that involves the use of autologous periosteal micrografts, which were used for a maxillary sinus floor lift in a 52-year-old female patient. This method allows for harvesting of a graft that is to be seeded on a PLGA scaffold and involves collection of a very little amount of palatal periosteal tissue in the same surgical site after elevation of a flap and disaggregation of it by using a Rigenera® filter. Histological samples collected at the time of implant installation demonstrate a good degree of bone regeneration. The clinical and radiographic outcomes at the 3-year follow-up visit showed an adequate stability of hard and soft tissues around the implants. This report demonstrates the possibility to obtain a sufficient quality and quantity of bone with a progenitor cell-based micrograft and in turn make the site appropriate for an implant-supported rehabilitation procedure, with stable results over a period of two years.
Collapse
|
148
|
Abstract
Mesenchymal stem cells (MSCs) have been discovered in almost every organ and tissue. MSCs are a heterogeneous population of cells with the capacity to self-renew and show multilineage differentiation. MSCs possess immunomodulatory properties by regulating multiple types of immune cells. They are emerging as a promising therapeutic agent, and have been widely used for cell-based tissue regeneration and immune therapies. A further understanding of the biological characteristics of MSCs is a prerequisite to develop more efficient MSC-based therapies. This article reviews the current understanding of different MSC populations in orofacial tissue compared with those derived from bone marrow.
Collapse
Affiliation(s)
- Xueli Mao
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Operative Dentistry and Endodontics, Guanghua School of Stomatology, Affiliated Stomatological Hospital, Sun Yat-sen University, 55 West Lingyuan Rd, Yuexiu District, Guangzhou 510055, China
| | - Yao Liu
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pediatric Dentistry, School of Stomatology, China Medical University, 117 South Nanjing Street, Heping District, Shenyang 110002, China
| | - Chider Chen
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Songtao Shi
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
149
|
Bone Morphogenetic Protein 6 Inhibits the Immunomodulatory Property of BMMSCs via Id1 in Sjögren's Syndrome. Stem Cells Int 2018; 2018:9837035. [PMID: 30174696 PMCID: PMC6098892 DOI: 10.1155/2018/9837035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/19/2018] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) treatment has emerged as a promising approach for treating Sjögren's syndrome (SS). Impaired immunoregulatory activities of bone marrow mesenchymal stem cells (BMMSCs) are found in both SS patients and animal models, and the underlying mechanism is poorly understood. Increased expression of BMP6 is reported to be related to SS. The aim herein was to determine the effects of BMP6 on BMMSCs function. BMMSCs were isolated from SS patients and NOD mice and showed a high level of BMP6 expression. The effects of BMP6 on BMMSCs function were investigated using in vitro BMMSCs differentiation and in vitro and in vivo T cell proliferation and polarization assays. BMP6 increased osteogenic differentiation of BMMSCs and inhibited the immunomodulatory properties of BMMSCs. BMP6 enhanced T cell proliferation and Th1/Th17 differentiation in a T cell-BMMSC coculture system. Mechanistically, BMP6 downregulated PGE2 and upregulated IFN-gamma via Id1 (inhibitor of DNA-binding protein 1). Neutralizing BMP6 and knockdown of Id1 could restore the BMMSCs immunosuppressive function both in vitro and in vivo. The present results suggest a novel role of Id1 in BMP-mediated MSCs function, which may contribute to a better understanding of the mechanism of action of MSCs in treating autoimmune diseases.
Collapse
|
150
|
Wang S, Zhu R, Li H, Li J, Han Q, Zhao RC. Mesenchymal stem cells and immune disorders: from basic science to clinical transition. Front Med 2018; 13:138-151. [PMID: 30062557 DOI: 10.1007/s11684-018-0627-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 02/08/2023]
Abstract
As a promising candidate seed cell type in regenerative medicine, mesenchymal stem cells (MSCs) have attracted considerable attention. The unique capacity of MSCs to exert a regulatory effect on immunity in an autologous/allergenic manner makes them an attractive therapeutic cell type for immune disorders. In this review, we discussed the current knowledge of and advances in MSCs, including its basic biological properties, i.e., multilineage differentiation, secretome, and immunomodulation. Specifically, on the basis of our previous work, we proposed three new concepts of MSCs, i.e., "subtotipotent stem cell" hypothesis, MSC system, and "Yin and Yang" balance of MSC regulation, which may bring new insights into our understanding of MSCs. Furthermore, we analyzed data from the Clinical Trials database ( http://clinicaltrials.gov ) on registered clinical trials using MSCs to treat a variety of immune diseases, such as graft-versus-host disease, systemic lupus erythematosus, and multiple sclerosis. In addition, we highlighted MSC clinical trials in China and discussed the challenges and future directions in the field of MSC clinical application.
Collapse
Affiliation(s)
- Shihua Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Rongjia Zhu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Hongling Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jing Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Qin Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China. .,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China. .,Peking Union Medical College Hospital, Beijing, 100005, China. .,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|